Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Pharmaceuticals (Basel) ; 17(5)2024 May 11.
Article in English | MEDLINE | ID: mdl-38794191

ABSTRACT

Gastrin-releasing peptide receptor (GRPR) is overexpressed in various cancers and is a promising target for cancer diagnosis and therapy. However, the high pancreas uptake and/or metabolic instability observed for most reported GRPR-targeted radioligands might limit their clinical applications. Our group recently reported a GRPR-targeted antagonist tracer, [68Ga]Ga-TacsBOMB2 ([68Ga]Ga-DOTA-Pip-D-Phe6-Gln7-Trp8-Ala9-Val10-Gly11-His12-Leu13ψThz14-NH2), which showed a minimal pancreas uptake in a preclinical mouse model. In this study, we synthesized four derivatives with unnatural amino acid substitutions (Tle10-derived Ga-LW01158, NMe-His12-derived Ga-LW01160, α-Me-Trp8- and Tle10-derived Ga-LW01186, and Tle10- and N-Me-Gly11-derived Ga-LW02002) and evaluated their potential for detecting GRPR-expressing tumors with positron emission tomography (PET). The binding affinities (Ki(GRPR)) of Ga-LW01158, Ga-LW01160, Ga-LW01186, and Ga-LW02002 were 5.11 ± 0.47, 187 ± 17.8, 6.94 ± 0.95, and 11.0 ± 0.39 nM, respectively. [68Ga]Ga-LW01158, [68Ga]Ga-LW01186, and [68Ga]Ga-LW02002 enabled clear visualization of subcutaneously implanted human prostate cancer PC-3 tumor xenografts in mice in PET images. Ex vivo biodistribution studies showed that [68Ga]Ga-LW01158 had the highest tumor uptake (11.2 ± 0.65 %ID/g) and good tumor-to-background uptake ratios at 1 h post-injection. Comparable in vivo stabilities were observed for [68Ga]Ga-LW01158, [68Ga]Ga-LW01186, and [68Ga]Ga-LW02002 (76.5-80.7% remaining intact in mouse plasma at 15 min post-injection). In summary, the Tle10 substitution, either alone or combined with α-Me-Trp8 or NMe-Gly11 substitution, in Ga-TacsBOMB2 generates derivatives that retained good GRPR binding affinity and in vivo stability. With good tumor uptake and tumor-to-background imaging contrast, [68Ga]Ga-LW01158 is promising for detecting GRPR-expressing lesions with PET.

2.
J Transl Med ; 22(1): 378, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649894

ABSTRACT

BACKGROUND: Bacteria-based cancer therapy have demonstrated innovative strategies to combat tumors. Recent studies have focused on gram-negative bacterial outer membrane vesicles (OMVs) as a novel cancer immunotherapy strategy due to its intrinsic properties as a versatile carrier. METHOD: Here, we developed an Human Papillomavirus (HPV)-associated E7 antigen displaying Salmonella-derived OMV vaccine, utilizing a Poly(L-arginine) cell penetrating peptide (CPP) to enhance HPV16 E7 (aa49-67) H-2 Db and OMV affinity, termed SOMV-9RE7. RESULTS: Due to OMV's intrinsic immunogenic properties, SOMV-9RE7 effectively activates adaptive immunity through antigen-presenting cell uptake and antigen cross-presentation. Vaccination of engineered OMVs shows immediate tumor suppression and recruitment of infiltrating tumor-reactive immune cells. CONCLUSION: The simplicity of the arginine coating strategy boasts the versatility of immuno-stimulating OMVs that can be broadly implemented to personalized bacterial immunotherapeutic applications.


Subject(s)
Arginine , Cancer Vaccines , Papillomavirus E7 Proteins , Papillomavirus E7 Proteins/immunology , Cancer Vaccines/immunology , Humans , Animals , Bacterial Outer Membrane/immunology , Mice, Inbred C57BL , Female
3.
Eur J Med Chem ; 268: 116238, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38367492

ABSTRACT

Fibroblast activation protein-α (FAP) is a marker of cancer-associated fibroblasts (CAFs) that constitute a significant portion of most carcinomas. Since it plays a critical role in tumor growth and metastasis, its timely detection to identify tumor lesions in early developmental stages using targeted radiopharmaceuticals has gained significant impetus. In the present work, two novel FAP-targeted precursors SB03178 and SB04033 comprising of an atypical benzo[h]quinoline construct were synthesized and either chelated to diagnostic radionuclide gallium-68 or therapeutic radionuclide lutetium-177, with ≥90% radiochemical purities and 22-76% decay-corrected radiochemical yields. natGa-labeled complexes displayed dose-dependent FAP inhibition, with binding potency of natGa-SB03178 being ∼17 times higher than natGa-SB04033. To evaluate their pharmacokinetic profiles, PET imaging and ex vivo biodistribution analyses were executed in FAP-overexpressing HEK293T:hFAP tumor-bearing mice. While both tracers displayed clear tumor visualization that was primarily FAP-arbitrated, with negligible uptake in most peripheral tissues, [68Ga]Ga-SB03178 demonstrated higher tumor uptake and superior tumor-to-background contrast ratios than [68Ga]Ga-SB04033. 177Lu-labeled SB03178 was subjected to tumor retention studies, mouse dosimetry profiling and mouse-to-human dose extrapolations also using the HEK293T:hFAP tumor model. [177Lu]Lu-SB03178 exhibited a combination of high and sustained tumor uptake, with excellent tumor-to-critical organ uptake ratios resulting in a high radiation absorbed dose to the tumor and a low estimated whole-body dose to humans. Our preliminary findings are considerably encouraging to support clinical development of [68Ga]Ga-/[177Lu]Lu-SB03178 theranostic pair for use in a vast majority of FAP-overexpressing neoplasms, particularly carcinomas.


Subject(s)
Carcinoma , Endopeptidases , Membrane Proteins , Quinolines , Humans , Animals , Mice , Gallium Radioisotopes , Tissue Distribution , HEK293 Cells , Radioisotopes , Radiopharmaceuticals/pharmacokinetics , Quinolines/chemistry , Positron Emission Tomography Computed Tomography/methods , Cell Line, Tumor
4.
EJNMMI Radiopharm Chem ; 9(1): 8, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38305955

ABSTRACT

BACKGROUND: Overexpressed in various solid tumors, gastrin-releasing peptide receptor (GRPR) is a promising cancer imaging marker and therapeutic target. Although antagonists are preferable for the development of GRPR-targeted radiopharmaceuticals due to potentially fewer side effects, internalization of agonists may lead to longer tumor retention and better treatment efficacy. In this study, we systematically investigated unnatural amino acid substitutions to improve in vivo stability and tumor uptake of a previously reported GRPR-targeted agonist tracer, [68Ga]Ga-TacBOMB2 (68Ga-DOTA-Pip-D-Phe6-Gln7-Trp8-Ala9-Val10-Gly11-His12-Leu13-Thz14-NH2). RESULTS: Unnatural amino acid substitutions were conducted for Gln7, Trp8, Ala9, Val10, Gly11 and His12, either alone or in combination. Out of 25 unnatural amino acid substitutions, tert-Leu10 (Tle10) and NMe-His12 substitutions were identified to be preferable modifications especially in combination. Compared with the previously reported [68Ga]Ga-TacBOMB2, the Tle10 and NMe-His12 derived [68Ga]Ga-LW01110 showed retained agonist characteristics and improved GRPR binding affinity (Ki = 7.62 vs 1.39 nM), in vivo stability (12.7 vs 89.0% intact tracer in mouse plasma at 15 min post-injection) and tumor uptake (5.95 vs 16.6 %ID/g at 1 h post-injection). CONCLUSIONS: Unnatural amino acid substitution is an effective strategy to improve in vivo stability and tumor uptake of peptide-based radiopharmaceuticals. With excellent tumor uptake and tumor-to-background contrast, [68Ga]Ga-LW01110 is promising for detecting GRPR-expressing cancer lesions with PET. Since agonists can lead to internalization upon binding to receptors and foreseeable long tumor retention, our optimized GRPR-targeted sequence, [Tle10,NMe-His12,Thz14]Bombesin(7-14), is a promising template for use for the design of GRPR-targeted radiotherapeutic agents.

5.
Molecules ; 29(4)2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38398552

ABSTRACT

Some bispecific radiotracers have been developed to overcome the limitations of monospecific tracers and improve detection sensitivity for heterogeneous tumor lesions. Here, we aim to synthesize two bispecific tracers targeting prostate-specific membrane antigen (PSMA) and fibroblast activation protein (FAP), which are key markers expressed in prostate cancer. A pyridine-based FAP-targeted ligand was synthesized through multi-step organic synthesis and then connected to the 2-Nal-containing PSMA-targeted motif. The Ki(PSMA) values of Ga-complexed bispecific ligands, Ga-AV01084 and Ga-AV01088, were 11.6 ± 3.25 and 28.7 ± 6.05 nM, respectively, and the IC50(FAP) values of Ga-AV01084 and Ga-AV01088 were 10.9 ± 0.67 and 16.7 ± 1.53 nM, respectively. Both [68Ga]Ga-AV01084 and [68Ga]Ga-AV01088 enabled the visualization of PSMA-expressing LNCaP tumor xenografts and FAP-expressing HEK293T:hFAP tumor xenografts in PET images acquired at 1 h post-injection. However, the tumor uptake values from the bispecific tracers were still lower than those obtained from the monospecific tracers, PSMA-targeted [68Ga]Ga-PSMA-617 and FAP-targeted [68Ga]Ga-AV02070. Further investigations are needed to optimize the selection of linkers and targeted pharmacophores to improve the tumor uptake of bispecific PSMA/FAP tracers for prostate cancer imaging.


Subject(s)
Gallium Radioisotopes , Prostatic Neoplasms , Male , Humans , HEK293 Cells , Pharmacophore , Radiopharmaceuticals/metabolism , Prostatic Neoplasms/pathology , Pyridines , Positron-Emission Tomography , Cell Line, Tumor
6.
Oncoimmunology ; 13(1): 2298444, 2024.
Article in English | MEDLINE | ID: mdl-38170154

ABSTRACT

Bacteria-based cancer therapy employs various strategies to combat tumors, one of which is delivering tumor-associated antigen (TAA) to generate specific immunity. Here, we utilized a poly-arginine extended HPV E7 antigen (9RE7) for attachment on Salmonella SL7207 outer membrane to synthesize the bacterial vaccine Salmonella-9RE7 (Sal-9RE7), which yielded a significant improvement in the amount of antigen presentation compared to the previous lysine-extended antigen coating strategy. In TC-1 tumor mouse models, Sal-9RE7 monotherapy decreased tumor growth by inducing E7 antigen-specific immunity. In addition, pairing Sal-9RE7 with adjuvant Albumin-IFNß (Alb-IFNß), a protein cytokine fusion, the combination significantly increased the antitumor efficacy and enhanced immunogenicity in the tumor microenvironment (TME). Our study made a significant contribution to personalized bacterial immunotherapy via TAA delivery and demonstrated the advantage of combination therapy.


Subject(s)
Interferon Type I , Neoplasms , Animals , Mice , Papillomavirus E7 Proteins/genetics , CD8-Positive T-Lymphocytes , Neoplasms/therapy , Antigens, Neoplasm , Immunotherapy , Salmonella , Tumor Microenvironment
7.
bioRxiv ; 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37398082

ABSTRACT

The Ras/PI3K/ERK signaling network is frequently mutated in various human cancers including cervical cancer and pancreatic cancer. Previous studies showed that the Ras/PI3K/ERK signaling network displays features of excitable systems including propagation of activity waves, all-or-none responses, and refractoriness. Oncogenic mutations lead to enhanced excitability of the network. A positive feedback loop between Ras, PI3K, the cytoskeleton, and FAK was identified as a driver of excitability. In this study, we investigated the effectiveness of targeting signaling excitability by inhibiting both FAK and PI3K in cervical and pancreatic cancer cells. We found that the combination of FAK and PI3K inhibitors synergistically suppressed the growth of select cervical and pancreatic cancer cell lines through increased apoptosis and decreased mitosis. In particular, FAK inhibition caused downregulation of PI3K and ERK signaling in cervical cancer but not pancreatic cancer cells. Interestingly, PI3K inhibitors activated multiple receptor tyrosine kinases (RTKs), including insulin receptor and IGF-1R in cervical cancer cells, as well as EGFR, Her2, Her3, Axl, and EphA2 in pancreatic cancer cells. Our results highlight the potential of combining FAK and PI3K inhibition for treating cervical and pancreatic cancer, although appropriate biomarkers for drug sensitivity are needed, and concurrent targeting of RTKs may be required for resistant cells.

8.
STAR Protoc ; 3(3): 101611, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36042884

ABSTRACT

We recently developed a biosensor barcoding approach for highly multiplexed tracking of molecular activities in live cells. In this protocol, we detail the labeling of cells expressing different genetically encoded fluorescent biosensors with a pair of barcoding proteins and parallel imaging. Signals from cells with the same barcodes are then pooled together to obtain the dynamics of the corresponding biosensor activity. We describe the steps involved in cell barcoding, image acquisition, and analysis by deep learning models. For complete details on the use and execution of this protocol, please refer to Yang et al. (2021).


Subject(s)
Biosensing Techniques , Biosensing Techniques/methods , Proteins
9.
Int J Nanomedicine ; 16: 7813-7830, 2021.
Article in English | MEDLINE | ID: mdl-34880610

ABSTRACT

INTRODUCTION: Osteoporosis is a result of an imbalance in bone remodeling. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have been considered as a potentially promising treatment for osteoporosis. However, the therapeutic effect, genetic alterations, and in vivo behavior of exogenous EVs for osteoporosis in mice models remain poorly understood. METHODS: A multiplexed molecular imaging strategy was constructed by micro-positron emission tomography (µPET)/computed tomography (CT), µCT, and optical imaging modality which reflected the osteoblastic activity, microstructure, and in vivo behavior of EVs, respectively. RNA sequencing was used to analyze the cargo of EVs, and the bone tissues of ovariectomized (OVX) mice post EV treatment. RESULTS: The result of [18F]NaF µPET showed an increase in osteoblastic activity in the distal femur of EV-treated mice, and the bone structural parameters derived from µCT were also improved. In terms of in vivo behavior of exogenous EVs, fluorescent dye-labeled EVs could target the distal femur of mice, whereas the uptakes of bone tissues were not significantly different between OVX mice and healthy mice. RNA sequencing demonstrated upregulation of ECM-related genes, which might associate with the PI3K/AKT signaling pathway, in line with the results of microRNA analysis showing that mir-21, mir-29, mir-221, and let-7a were enriched in Wharton's jelly-MSC-EVs and correlated to the BMP and PI3K/AKT signaling pathways. CONCLUSION: The therapeutic effect of exogenous WJ-MSC-EVs in the treatment of osteoporosis was successfully assessed by a multiplexed molecular imaging strategy. The RNA sequencing demonstrated the possible molecular targets in the regulation of bone remodeling. The results highlight the novelty of diagnostic and therapeutic strategies of EV-based treatment for osteoporosis.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , MicroRNAs , Osteoporosis , Wharton Jelly , Animals , Mice , Molecular Imaging , Osteoporosis/diagnostic imaging , Osteoporosis/therapy , Phosphatidylinositol 3-Kinases , Sequence Analysis, RNA
10.
Mol Imaging Biol ; 23(3): 361-371, 2021 06.
Article in English | MEDLINE | ID: mdl-33216285

ABSTRACT

PURPOSE: Mesenchymal stem cell-derived EVs (MSC-EVs) are demonstrated to have similar therapeutic effect as their cells of origin and represent an attractive cell-free stem cell therapy. With the potential to be the future medical regimen, the information of fate and behavior of MSC-EVs in the living subject should be urgently gathered. This study aimed to track MSC-EVs by 111In-labeling and µSPECT/CT imaging. PROCEDURES: Wharton's jelly-MSC-EVs (WJ-MSC-EVs) were isolated using Exo-Prep kit followed by characterization of expressing markers and size. After labeled by 111In-oxine, 111In-EVs were injected into C57BL/6 mice followed by µSPECT/CT imaging. Organs were then taken out for ex vivo biodistribution analysis. RESULTS: The radiochemical purity of 111In-EVs was > 90 % and remained stable up to 24 h. The image results showed that with injection of 111In-EVs, the signal mainly accumulated in the liver, spleen, and kidney, compared to that in lung and kidney after 111In-oxine injection. The ex vivo biodistribution showed the similar pattern to that of imaging. Chelation of free 111In with EDTA was found necessary to reduce the nonspecific accumulation of signal. CONCLUSION: This study demonstrated the feasibility of radiolabeling WJ-MSC-EVs with 111In-oxine for in vivo imaging and quantitative analysis in a mouse model. This simple and quick labeling method preserves the characteristics of WJ-MSC-EVs. The results in this study provide a thorough and objective basis for future clinical study.


Subject(s)
Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/cytology , Organometallic Compounds/chemistry , Oxyquinoline/analogs & derivatives , Tomography, Emission-Computed, Single-Photon/methods , Tomography, X-Ray Computed/methods , Animals , Cell Lineage , Cell Proliferation , Culture Media, Conditioned , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Nanoparticles , Oxyquinoline/chemistry , Tissue Distribution , Wharton Jelly
11.
Int J Mol Sci ; 21(18)2020 Sep 09.
Article in English | MEDLINE | ID: mdl-32916962

ABSTRACT

Malignant melanoma is the most harmful type of skin cancer and its incidence has increased in this past decade. Early diagnosis and treatment are urgently desired. In this study, we conjugated picolinamide/nicotinamide with the pharmacophore of 131I-MIP-1145 to develop 131I-iodofluoropicolinamide benzamide (131I-IFPABZA) and 131I-iodofluoronicotiamide benzamide (131I-IFNABZA) with acceptable radiochemical yield (40 ± 5%) and high radiochemical purity (>98%). We also presented their biological characteristics in melanoma-bearing mouse models. 131I-IFPABZA (Log P = 2.01) was more lipophilic than 131I-IFNABZA (Log P = 1.49). B16F10-bearing mice injected with 131I-IFNABZA exhibited higher tumor-to-muscle ratio (T/M) than those administered with 131I-IFPABZA in planar γ-imaging and biodistribution studies. However, the imaging of 131I-IFNABZA- and 131I-IFPABZA-injected mice only showed marginal tumor uptake in A375 amelanotic melanoma-bearing mice throughout the experiment period, indicating the high binding affinity of these two radiotracers to melanin. Comparing the radiation-absorbed dose of 131I-IFNABZA with the melanin-targeted agents reported in the literature, 131I-IFNABZA exerts lower doses to normal tissues on the basis of similar tumor dose. Based on the in vitro and in vivo studies, we clearly demonstrated the potential of using 131I-IFNABZA as a theranostic agent against melanoma.


Subject(s)
Benzamides/therapeutic use , Iodine Radioisotopes/therapeutic use , Melanoma, Experimental/therapy , Skin Neoplasms/therapy , Animals , Benzamides/chemistry , Cell Line, Tumor , Humans , Iodine Radioisotopes/chemistry , Melanins/metabolism , Melanoma, Experimental/diagnostic imaging , Mice, Inbred C57BL , Niacinamide/chemistry , Picolinic Acids/chemistry , Precision Medicine , Radionuclide Imaging , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/metabolism , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Skin Neoplasms/diagnostic imaging , Xenograft Model Antitumor Assays
12.
J Mater Chem B ; 8(1): 65-77, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31768514

ABSTRACT

Gold nanostars (AuNSs), with unique physicochemical properties, are thought to be a promising agent for photothermal therapy (PTT). In this study, we prepared PEGylated gold nanostars (pAuNSs) using the HEPES-reduction method. The high photothermal conversion efficiency (∼80%) and photothermal stability of pAuNSs were demonstrated in vitro and in vivo. 111In-DTPA-pAuNSs were prepared as a radioactive surrogate for the biodistribution studies of pAuNSs. In both microSPECT/CT images and the biodistribution study, the tumor-to-muscle (T/M) ratio reached a maximum at 24 h post intravenous injection of 111In-DTPA-pAuNSs. The high linear correlation between the 111In radioactivity and the gold content in the tumors (R2 0.86-0.99) indicated that 111In-DTPA-pAuNSs were appropriate for noninvasively tracking pAuNSs in vivo after systemic administration. Histological examination after silver enhancement staining clearly illustrated that the accumulated pAuNSs in the tumors were mainly located on the luminal surface of vessels. The mice bearing a SKOV-3 xenograft exhibited remarkable therapeutic efficacy with negligible organ damage after receiving pAuNS-mediated photothermal therapy. Our findings suggested that pAuNSs, together with their radioactive surrogate 111In-DTPA-pAuNSs, are promising for applications in image-guided photothermal therapy.


Subject(s)
Gold/pharmacokinetics , Metal Nanoparticles/therapeutic use , Neoplasms/therapy , Phototherapy/methods , Polyethylene Glycols/pharmacokinetics , Theranostic Nanomedicine/methods , Animals , Cell Line, Tumor , Female , Gold/therapeutic use , Humans , Mice , Mice, Inbred BALB C
13.
Int J Mol Sci ; 20(1)2019 Jan 08.
Article in English | MEDLINE | ID: mdl-30626093

ABSTRACT

Colorectal cancer is one of the major causes of cancer-related death in Taiwan and worldwide. Patients with peritoneal metastasis from colorectal cancer have reduced overall survival and poor prognosis. Hybrid protein-inorganic nanoparticle systems have displayed multifunctional applications in solid cancer theranostics. In this study, a gold nanocore-encapsulated human serum albumin nanoparticle (Au@HSANP), which is a hybrid protein-inorganic nanoparticle, and its radioactive surrogate 111In-labeled Au@HSANP (111In-Au@HSANP), were developed and their biological behaviors were investigated in a tumor/ascites mouse model. 111In-Au@HSANP was injected either intravenously (iv) or intraperitoneally (ip) in CT-26 tumor/ascites-bearing mice. After ip injection, a remarkable and sustained radioactivity retention in the abdomen was noticed, based on microSPECT images. After iv injection, however, most of the radioactivity was accumulated in the mononuclear phagocyte system. The results of biodistribution indicated that ip administration was significantly more effective in increasing intraperitoneal concentration and tumor accumulation than iv administration. The ratios of area under the curve (AUC) of the ascites and tumors in the ip-injected group to those in the iv-injected group was 93 and 20, respectively. This study demonstrated that the ip injection route would be a better approach than iv injections for applying gold-albumin nanoparticle in peritoneal metastasis treatment.


Subject(s)
Ascites/pathology , Gold/administration & dosage , Nanoparticles/administration & dosage , Serum Albumin, Human/administration & dosage , Administration, Intravenous , Animals , Area Under Curve , Cell Survival , Disease Models, Animal , Dynamic Light Scattering , Indium Radioisotopes/chemistry , Indium Radioisotopes/pharmacokinetics , Injections, Intraperitoneal , Injections, Intravenous , Mice , Nanoparticles/ultrastructure , Particle Size , Serum Albumin, Human/pharmacokinetics , Tissue Distribution , Tomography, Emission-Computed, Single-Photon
14.
ACS Appl Bio Mater ; 2(1): 533-543, 2019 Jan 22.
Article in English | MEDLINE | ID: mdl-35016317

ABSTRACT

A combined photothermal therapy (PTT) and chemotherapy (chemo) were performed in vitro on B16F10 melanoma cells and in vivo using melanoma bearing C57BL/6 mice. The 785 nm (100 mW) irradiated gold nanorods (AuNRs) were used as the PT agent, and electrostatically conjugated Doxorubicin (Dox) to a nanocarrier graphene oxide (GO) worked as the chemotherapeutic. Selection of dosage was optimized from the individual viability studies, and finally a combined therapeutic (AuNR (100 ppm), GO (125, and 250 ppm), Dox (0.0058, and 0.00058 ppm)), was delivered in vitro. PTT, followed by chemo, sequentially, resulted in <10% viability, whereas simultaneous PTT with chemo resulted in a viability of ∼40% for the melanoma cells. Flow cytometry indicated optical inhomogeneity in the cells that internalized GO, and AuNR; however, the Dox amount was identical within the cells treated with or without PTT. Confocal microscopy revealed that GO+Dox was internalized, and Dox was distributed uniformly within the cells irrespective of the treatment protocol. In vivo results in melanoma bearing C57BL/6 mice resembled the in vitro data closely. The tumor growth inhibition index was highest at 0.78 for the group receiving sequential treatment, followed by 0.61 for those receiving simultaneous treatment, where the control group had a score of 0. For the sequential treatment, presoftening of the cells with PTT, followed by the chemo resulted in significantly improved toxicity of the treatment, whereas simultaneous PTT with chemo results were dominated by the Dox alone.

15.
ACS Appl Bio Mater ; 2(8): 3573-3581, 2019 Aug 19.
Article in English | MEDLINE | ID: mdl-35030743

ABSTRACT

The efficacy of gold nanoparticle (AuNP)-assisted radiofrequency (RF)-induced hyperthermia employing the Kanzius device remains controversial. Different from the Kanzius device, modulated electro-hyperthermia (mEHT) utilizes the capacitive-impedance coupled 13.56 MHz radiofrequency (RF) current and has been approved for clinical cancer treatment. In this study, we investigated the heating characteristics of spherical-, urchin-, and rod-like AuNPs of a similar 50 nm size upon exposure to a 13.56 MHz radiofrequency using the LabEHY-105CL, an in vivo mEHT device. We found that, regardless of the AuNPs' sphere-, urchin- or rod-like shape, purified gold nanoparticle solution would not promote heat generation. The temperature elevation during radiofrequency irradiation was solely attributed to the ionic background of the solution. The AuNPs present in the medium (≤25 ppm) showed no effect on selective cell killing of malignant cells, whereas the AuNPs incorporated in the cells diminished the cell selectivity as well as cell death and acted as protectors in mEHT cancer treatment. Our study suggested that (1) the temperature elevation induced by 50 nm AuNPs in the 13.56 MHz radiofrequency field was negligible and was shape-independent, and (2) the presence of AuNPs would alter the cell-killing effect of modulated electro-hyperthermia.

16.
Cell Death Discov ; 4: 100, 2018.
Article in English | MEDLINE | ID: mdl-30393570

ABSTRACT

Human head and neck squamous cell carcinoma (HNSCC) is usually treated with chemoradiotherapy, but the therapeutic efficacy could be hampered by intrinsic radioresistance and early relapse. Repeated administrations of rhenium-188 (188Re)-conjugated radiopharmaceutical has been reported to escalate the radiation doses for better control of advanced human cancers. Here we found that high dosage of 188Re-liposome, the liposome-encapsulated 188Re nanoparticles exhibited significant killing effects on HNSCC FaDu cells and SAS cells but not on OECM-1 cells. To investigate the biological and pharmaceutical responses of high 188Re-liposomal dosage in vivo, repeated doses of 188Re-liposome was injected into the orthotopic tumor model. FaDu cells harboring luciferase reporter genes were implanted in the buccal positions of nude mice followed by intravenous injection of 188Re-liposome. The Cerenkov luminescence imaging (CLI) was performed to demonstrate an increased accumulation of 188Re-liposome in the tumor lesion of nude mice with repeated doses compared to a single dose. Repeated doses also enhanced tumor growth delay and elongated the survival of tumor-bearing mice. These observations were associated with significant loss of Ki-67 proliferative marker and epithelial-mesenchymal transition (EMT) markers in excised tumor cells. The body weights of mice were not significantly changed using different doses of 188Re-liposome, yet repeated doses led to lower blood counts than a single dose. Furthermore, the pharmacokinetic analysis showed that the internal circulation of repeated 188Re-liposomal therapy was elongated. The biodistribution analysis also demonstrated that accumulations of 188Re-liposome in tumor lesions and bone marrow were increased using repeated doses. The absorbed dose of repeated doses over a single dose was about twofold estimated for a 1 g tumor. Together, these data suggest that the radiopharmacotherapy of 188Re-liposome can enhance tumor suppression, survival extension, and internal circulation without acute toxicity using repeated administrations.

17.
Theranostics ; 8(11): 3164-3175, 2018.
Article in English | MEDLINE | ID: mdl-29896310

ABSTRACT

Rationale: Increasing frequency of human exposure to PEG-related products means that healthy people are likely to have pre-existing anti-PEG antibodies (pre-αPEG Ab). However, the influence of pre-αPEG Abs on the pharmacokinetics (PK) and therapeutic efficacy of LipoDox is unknown. Methods: We generated two pre-αPEG Ab mouse models. First, naïve mice were immunized with PEGylated protein to generate an endogenous αPEG Ab titer (endo αPEG). Second, monoclonal αPEG Abs were passively transferred (αPEG-PT) into naïve mice to establish a αPEG titer. The naïve, endo αPEG and αPEG-PT mice were intravenously injected with 111in-labeled LipoDox to evaluate its PK. Tumor-bearing naïve, endo αPEG and αPEG-PT mice were intravenously injected with 111in-labeled LipoDox to evaluate its biodistribution. The therapeutic efficacy of LipoDox was estimated in the tumor-bearing mice. Results: The areas under the curve (AUC)last of LipoDox in endo αPEG and αPEG-PT mice were 11.5- and 15.6- fold less, respectively, than that of the naïve group. The biodistribution results suggested that pre-αPEG Ab can significantly reduce tumor accumulation and accelerate blood clearance of 111In-labeled LipoDox from the spleen. The tumor volumes of the tumor-bearing endo αPEG and αPEG-PT mice after treatment with LipoDox were significantly increased as compared with that of the tumor-bearing naïve mice. Conclusions: Pre-αPEG Abs were found to dramatically alter the PK and reduce the tumor accumulation and therapeutic efficacy of LipoDox. Pre-αPEG may have potential as a marker to aid development of personalized therapy using LipoDox and achieve optimal therapeutic efficacy.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Antibodies/immunology , Doxorubicin/analogs & derivatives , Neoplasms, Experimental/drug therapy , Animals , Antibiotics, Antineoplastic/immunology , Antibiotics, Antineoplastic/pharmacokinetics , Antibodies/blood , Doxorubicin/immunology , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Female , Liposomes/pharmacokinetics , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/therapeutic use
18.
Nanotechnology ; 25(29): 295102, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24990295

ABSTRACT

Gold nanoparticles (AuNPs) are widely applied to the diagnosis and treatment of cancer and can be modified to contain target-specific ligands via gold-thiolate bonding. This study investigated the pharmacokinetics and microdistribution of antibody-mediated active targeting gold nanoparticles in mice with subcutaneous lung carcinoma. We conjugated AuNPs with cetuximab (C225), an antibody-targeting epidermal growth factor receptor (EGFR), and then labeled with In-111, which created EGFR-targeted AuNPs. In vitro studies showed that after a 2 h incubation, the uptake of C225-conjugated AuNPs in high EGFR-expression A549 cells was 14.9-fold higher than that of PEGylated AuNPs; furthermore, uptake was also higher at 3.8-fold when MCF7 cells with lower EGFR-expression were used. MicroSPECT/CT imaging and a biodistribution study conducted by using a A549 tumor xenograft mouse model provided evidence of elevated uptake of the C225-conjugated AuNPs into the tumor cells as a result of active targeting. Moreover, the microdistribution of PEGylated AuNPs revealed that a large portion of AuNPs remained in the tumor interstitium, whereas the C225-conjugated AuNPs displayed enhanced internalization via antibody-mediated endocytosis. Our findings suggest that the anti-EGFR antibody-conjugated AuNPs are likely to be a plausible nano-sized vehicle for drug delivery to EGFR-expressing tumors.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Carcinoma/drug therapy , Lung Neoplasms/drug therapy , Nanoconjugates/therapeutic use , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antineoplastic Agents/chemical synthesis , Cetuximab , Disease Models, Animal , Female , Gold/chemistry , Gold/pharmacokinetics , Mice , Mice, Inbred BALB C , Microspectrophotometry , Nanoconjugates/chemistry , Surface Plasmon Resonance , Tumor Cells, Cultured
19.
Bioorg Med Chem Lett ; 23(11): 3180-5, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23628334

ABSTRACT

This study evaluated the tumor targeting and therapeutic efficacy of a novel theranostic agent (131)I-labeled immuno-gold-nanoparticle ((131)I-C225-AuNPs-PEG) for high epidermal growth factor receptor (EGFR)-expressed A549 human lung cancer. Confocal microscopy demonstrated the specific uptake of C225-AuNPs-PEG in A549 cells. (131)I-C225-AuNPs-PEG induced a significant reduction in cell viability, which was not observed when incubated with AuNPs-PEG and C225-AuNPs-PEG. MicroSPECT/CT imaging of tumor-bearing mice after intravenous injection of (123)I-C225-AuNPs-PEG revealed significant radioactivity retention in tumor suggested that (131)I-labeled C225-conjugated radioimmuno-gold-nanoparticles may provide a new approach of targeted imaging and therapy towards high EGFR-expressed cancers.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Antineoplastic Agents/chemistry , ErbB Receptors/antagonists & inhibitors , Gold/chemistry , Metal Nanoparticles/chemistry , Radiopharmaceuticals/chemistry , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/toxicity , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Cetuximab , Disease Models, Animal , Drug Evaluation, Preclinical , ErbB Receptors/metabolism , Humans , Injections, Intravenous , Iodine Radioisotopes/chemistry , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/drug therapy , Mice , Microscopy, Confocal , Polyethylene Glycols/chemistry , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/toxicity , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Transplantation, Heterologous
20.
Dev Neuropsychol ; 34(1): 85-102, 2009.
Article in English | MEDLINE | ID: mdl-19142768

ABSTRACT

Emotion regulation is a critical component of healthy development, yet few studies examine neural correlates of emotion regulation in childhood. In the present study, we assessed whether children's neurophysiological responses to salient and socially significant emotional distracters-emotional faces-were related to broader emotion regulation capacities. Emotion regulation was measured as attention performance following emotional distracters and as maternal report of child emotional dysregulation. Electroencephalography was recorded while participants (15 children aged 5-9) performed an attention task. Scalp-recorded event related potentials (ERPs) were time-locked to emotional distracters (fearful, sad, and neutral faces) and reflected a range of rapid attentional and face processing operations (P1, N1, N170, and Nc). P1 latencies were faster whereas N1 amplitudes were reduced to fearful compared to sad faces. Larger P1 and Nc amplitudes to fearful and sad faces were correlated with more effective emotion regulation. Results are discussed in terms of mechanisms in emotion regulation and the use of ERPs to detect early risk for psychopathology and inform intervention efforts.


Subject(s)
Emotions/physiology , Evoked Potentials, Visual/physiology , Face , Facial Expression , Analysis of Variance , Attention/physiology , Brain Mapping , Child , Child, Preschool , Electroencephalography/methods , Electrooculography/methods , Female , Humans , Male , Neuropsychological Tests , Pattern Recognition, Visual/physiology , Photic Stimulation/methods , Reaction Time/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...